1
|
Brimmer S, Ji P, Birla RK, Heinle JS, Grande-Allen JK, Keswani SG. Development of Novel 3D Spheroids for Discrete Subaortic Stenosis. Cardiovasc Eng Technol 2024:10.1007/s13239-024-00746-x. [PMID: 39495395 DOI: 10.1007/s13239-024-00746-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/07/2024] [Indexed: 11/05/2024]
Abstract
In this study, we propose a new method for bioprinting 3D Spheroids to study complex congenital heart disease known as discrete subaortic stenosis (DSS). The bioprinter allows us to manipulate the extrusion pressure to change the size of the spheroids, and the alginate porosity increases in size over time. The spheroids are composed of human umbilical vein endothelial cells (HUVECs), and we demonstrated that pressure and time during the bioprinting process can modulate the diameter of the spheroids. In addition, we used Pluronic acid to maintain the shape and position of the spheroids. Characterization of HUVECs in the spheroids confirmed their uniform distribution and we demonstrated cell viability as a function of time. Compared to traditional 2D cell cultures, the 3D spheroids model provides more relevant physiological environments, making it valuable for drug testing and therapeutic applications.
Collapse
Affiliation(s)
- Sunita Brimmer
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, TX, USA
- Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA
- Division of Congenital Heart Surgery, Texas Children's Hospital, Houston, TX, USA
| | - Pengfei Ji
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, TX, USA
- Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA
- Division of Congenital Heart Surgery, Texas Children's Hospital, Houston, TX, USA
| | - Ravi K Birla
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, TX, USA
- Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA
- Division of Congenital Heart Surgery, Texas Children's Hospital, Houston, TX, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Houston, TX, USA
| | - Jeffrey S Heinle
- Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA
- Division of Congenital Heart Surgery, Texas Children's Hospital, Houston, TX, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Houston, TX, USA
| | | | - Sundeep G Keswani
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, TX, USA.
- Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA.
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
- Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Houston, TX, USA.
- Feigin Center C.450.06, Texas Children's Hospital, 1102 Bates Ave, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Baduini IR, Castro Vildosola JE, Kavehmoghaddam S, Kiliç F, Nadeem SA, Nizama JJ, Rowand MA, Annapureddy D, Bryan CA, Do LH, Hsiao S, Jonnalagadda SA, Kasturi A, Mandava N, Muppavaram S, Ramirez B, Siner A, Suoto CN, Tamajal N, Scoma ER, Da Costa RT, Solesio ME. Type 2 diabetes mellitus and neurodegenerative disorders: The mitochondrial connection. Pharmacol Res 2024; 209:107439. [PMID: 39357690 DOI: 10.1016/j.phrs.2024.107439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
The incidence of type 2 diabetes mellitus (T2DM) has increased in our society in recent decades as the population ages, and this trend is not expected to revert. This is the same for the incidence of the main neurodegenerative disorders, including the two most common ones, which are, Alzheimer's and Parkinson's disease. Currently, no pharmacological therapies have been developed to revert or cure any of these pathologies. Interestingly, in recent years, an increased number of studies have shown a high co-morbidity between T2DM and neurodegeneration, as well as some common molecular pathways that are affected in both types of diseases. For example, while the etiopathology of T2DM and neurodegenerative disorders is highly complex, mitochondrial dysfunction has been broadly described in the early steps of both diseases; accordingly, this dysfunction has emerged as a plausible molecular link between them. In fact, the prominent role played by mitochondria in the mammalian metabolism of glucose places the physiology of the organelle in a central position to regulate many cellular processes that are affected in both T2DM and neurodegenerative disorders. In this collaborative review, we critically describe the relationship between T2DM and neurodegeneration; making a special emphasis on the mitochondrial mechanisms that could link these diseases. A better understanding of the role of mitochondria on the etiopathology of T2DM and neurodegeneration could pave the way for the development of new pharmacological therapies focused on the regulation of the physiology of the organelle. These therapies could, ultimately, contribute to increase healthspan.
Collapse
Affiliation(s)
- Isabella R Baduini
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Jose E Castro Vildosola
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sheida Kavehmoghaddam
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Fatmanur Kiliç
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - S Aiman Nadeem
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Juan J Nizama
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Marietta A Rowand
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Dileep Annapureddy
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Chris-Ann Bryan
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Lisa H Do
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Samuel Hsiao
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sai A Jonnalagadda
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Akhila Kasturi
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nikhila Mandava
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Sachin Muppavaram
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Bryan Ramirez
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Aleece Siner
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Christina N Suoto
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nasira Tamajal
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Ernest R Scoma
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Renata T Da Costa
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Maria E Solesio
- Department of Biology and Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA.
| |
Collapse
|
3
|
D'Egidio F, Castelli V, d'Angelo M, Ammannito F, Quintiliani M, Cimini A. Brain incoming call from glia during neuroinflammation: Roles of extracellular vesicles. Neurobiol Dis 2024; 201:106663. [PMID: 39251030 DOI: 10.1016/j.nbd.2024.106663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
The functionality of the central nervous system (CNS) relies on the connection, integration, and the exchange of information among neural cells. The crosstalk among glial cells and neurons is pivotal for a series of neural functions, such as development of the nervous system, electric conduction, synaptic transmission, neural circuit establishment, and brain homeostasis. Glial cells are crucial players in the maintenance of brain functionality in physiological and disease conditions. Neuroinflammation is a common pathological process in various brain disorders, such as neurodegenerative diseases, and infections. Glial cells, including astrocytes, microglia, and oligodendrocytes, are the main mediators of neuroinflammation, as they can sense and respond to brain insults by releasing pro-inflammatory or anti-inflammatory factors. Recent evidence indicates that extracellular vesicles (EVs) are pivotal players in the intercellular communication that underlies physiological and pathological processes. In particular, glia-derived EVs play relevant roles in modulating neuroinflammation, either by promoting or inhibiting the activation of glial cells and neurons, or by facilitating the clearance or propagation of pathogenic proteins. The involvement of EVs in neurodegenerative diseases such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Multiple Sclerosis (MS)- which share hallmarks such as neuroinflammation and oxidative stress to DNA damage, alterations in neurotrophin levels, mitochondrial impairment, and altered protein dynamics- will be dissected, showing how EVs act as pivotal cell-cell mediators of toxic stimuli, thereby propagating degeneration and cell death signaling. Thus, this review focuses on the EVs secreted by microglia, astrocytes, oligodendrocytes and in neuroinflammatory conditions, emphasizing on their effects on neurons and on central nervous system functions, considering both their beneficial and detrimental effects.
Collapse
Affiliation(s)
- Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo".
| | - Fabrizio Ammannito
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Massimiliano Quintiliani
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| |
Collapse
|
4
|
Tian C, Cai H, Ao Z, Gu L, Li X, Niu VC, Bondesson M, Gu M, Mackie K, Guo F. Engineering human midbrain organoid microphysiological systems to model prenatal PFOS exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174478. [PMID: 38964381 PMCID: PMC11404128 DOI: 10.1016/j.scitotenv.2024.174478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 μM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.
Collapse
Affiliation(s)
- Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Xiang Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Vivian C Niu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States; Bloomington High School South, Bloomington, IN 47401, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH 45229, Cincinnati, United States; University of Cincinnati School of Medicine, OH 45229, Cincinnati, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States.
| |
Collapse
|
5
|
Neziri S, Köseoğlu AE, Deniz Köseoğlu G, Özgültekin B, Özgentürk NÖ. Animal models in neuroscience with alternative approaches: Evolutionary, biomedical, and ethical perspectives. Animal Model Exp Med 2024. [PMID: 39375824 DOI: 10.1002/ame2.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/07/2024] [Indexed: 10/09/2024] Open
Abstract
Animal models have been a crucial tool in neuroscience research for decades, providing insights into the biomedical and evolutionary mechanisms of the nervous system, disease, and behavior. However, their use has raised concerns on several ethical, clinical, and scientific considerations. The welfare of animals and the 3R principles (replacement, reduction, refinement) are the focus of the ethical concerns, targeting the importance of reducing the stress and suffering of these models. Several laws and guidelines are applied and developed to protect animal rights during experimenting. Concurrently, in the clinic and biomedical fields, discussions on the relevance of animal model findings on human organisms have increased. Latest data suggest that in a considerable amount of time the animal model results are not translatable in humans, costing time and money. Alternative methods, such as in vitro (cell culture, microscopy, organoids, and micro physiological systems) techniques and in silico (computational) modeling, have emerged as potential replacements for animal models, providing more accurate data in a minimized cost. By adopting alternative methods and promoting ethical considerations in research practices, we can achieve the 3R goals while upholding our responsibility to both humans and other animals. Our goal is to present a thorough review of animal models used in neuroscience from the biomedical, evolutionary, and ethical perspectives. The novelty of this research lies in integrating diverse points of views to provide an understanding of the advantages and disadvantages of animal models in neuroscience and in discussing potential alternative methods.
Collapse
Affiliation(s)
- Sabina Neziri
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| | | | | | - Buminhan Özgültekin
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acıbadem University, Istanbul, Turkey
| | - Nehir Özdemir Özgentürk
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Yıldız Technical University, Istanbul, Turkey
| |
Collapse
|
6
|
Wang X, Lu J, Huang Y, Liu X, Fang G, Yang CT, Guo Z. Editorial: Emerging bioanalytical techniques and therapies for human disease models. Front Bioeng Biotechnol 2024; 12:1453813. [PMID: 39386041 PMCID: PMC11461311 DOI: 10.3389/fbioe.2024.1453813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Affiliation(s)
- Xuerui Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinnuo Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yixiao Huang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinhao Liu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guocheng Fang
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore, Singapore
| | - Chih-Tsung Yang
- Future Industries Institute, University of South Australia, Adelaide, SA, Australia
| | - Zhaobin Guo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
7
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
8
|
Barnes CA, Permenter MR, Vogt JA, Chen K, Beach TG. Human Alzheimer's Disease ATN/ABC Staging Applied to Aging Rhesus Macaque Brains: Association With Cognition and MRI-Based Regional Gray Matter Volume. J Comp Neurol 2024; 532:e25670. [PMID: 39315417 PMCID: PMC11451939 DOI: 10.1002/cne.25670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
The brain changes of Alzheimer's disease (AD) include Abeta (Aβ) amyloid plaques ("A"), abnormally phosphorylated tau tangles ("T"), and neurodegeneration ("N"). These have been used to construct in vivo and postmortem diagnostic and staging classifications for evaluating the spectrum of AD in the "ATN" and "ABC" ("B" for Braak tau stage, "C" for Consortium to Establish a Registry for Alzheimer's Disease [CERAD] neuritic plaque density) systems. Another common AD feature involves cerebral amyloid angiopathy (CAA). We report the first experiment to examine relationships among cognition, brain distribution of amyloid plaques, CAA, tau/tangles, and magnetic resonance imaging (MRI)-determined volume changes (as a measure of "N") in the same group of behaviorally characterized nonhuman primates. Both ATN and ABC systems were applied to a group of 32 rhesus macaques aged between 7 and 33 years. When an immunohistochemical method for "T" and "B" was used, some monkeys were "triple positive" on ATN, with a maximum ABC status of A1B2C3. With silver or thioflavin S methods, however, all monkeys were classified as T-negative and B0, indicating the absence of mature neurofibrillary tangles (NFTs) and hence neuropathologically defined AD. Although monkeys at extremes of the ATN and ABC classifications, or with frequent CAA, had significantly lower scores on some cognitive tests, the lack of fully mature NFTs or dementia-consistent cognitive impairment indicates that fully developed AD may not occur in rhesus macaques. There were sex differences noted in the types of histopathology present, and only CAA was significantly related to gray matter volume.
Collapse
Affiliation(s)
- Carol A Barnes
- Departments of Psychology, Neurology and Neuroscience, Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Tucson, Arizona, USA
| | - Michele R Permenter
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Julie A Vogt
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Kewei Chen
- Arizona State University, Tempe, Arizona, USA
| | - Thomas G Beach
- Department of Neuroscience, Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
9
|
Marshall EM, Rashidi AS, van Gent M, Rockx B, Verjans GMGM. Neurovirulence of Usutu virus in human fetal organotypic brain slice cultures partially resembles Zika and West Nile virus. Sci Rep 2024; 14:20095. [PMID: 39209987 PMCID: PMC11362282 DOI: 10.1038/s41598-024-71050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Usutu (USUV), West Nile (WNV), and Zika virus (ZIKV) are neurotropic arthropod-borne viruses (arboviruses) that cause severe neurological disease in humans. However, USUV-associated neurological disease is rare, suggesting a block in entry to or infection of the brain. We determined the replication, cell tropism and neurovirulence of these arboviruses in human brain tissue using a well-characterized human fetal organotypic brain slice culture model. Furthermore, we assessed the efficacy of interferon-β and 2'C-methyl-cytidine, a synthetic nucleoside analogue, in restricting viral replication. All three arboviruses replicated within the brain slices, with WNV reaching the highest titers, and all primarily infected neuronal cells. USUV- and WNV-infected cells exhibited a shrunken morphology, not associated with detectable cell death. Pre-treatment with interferon-β inhibited replication of all arboviruses, while 2'C-methyl-cytidine reduced only USUV and ZIKV titers. Collectively, USUV can infect human brain tissue, showing similarities in tropism and neurovirulence as WNV and ZIKV. These data suggest that a blockade to infection of the human brain may not be the explanation for the low clinical incidence of USUV-associated neurological disease. However, USUV replicated more slowly and to lower titers than WNV, which could help to explain the reduced severity of neurological disease resulting from USUV infection.
Collapse
Affiliation(s)
- Eleanor M Marshall
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ahmad S Rashidi
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- HerpeslabNL of the Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michiel van Gent
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- HerpeslabNL of the Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
- HerpeslabNL of the Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Tokuoka SM, Hamano F, Kobayashi A, Adachi S, Andou T, Natsume T, Oda Y. Plasma proteomics and lipidomics facilitate elucidation of the link between Alzheimer's disease development and vessel wall fragility. Sci Rep 2024; 14:19901. [PMID: 39191863 DOI: 10.1038/s41598-024-71097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Proximity Extension Assay (PEA) and mass spectrometry (MS) methodologies were utilized for the proteomic and lipidomic characterization of plasma specimens from patients who developed Alzheimer's disease. Proteomics was performed by both PEA and Liquid Chromatography (LC)/MS in this study, but all the more because LC/MS generally tends to be biased towards proteins with high expression and high variability, generating hypotheses proved challenging. Consequently, attempt was made to interpret the results from the PEA data. There were 150 significantly variable proteins and 68 lipids among 1000 proteins and 400 lipids. Pathway analysis was performed for both total and variable proteins measured to reduce bias, and it appeared that vascular fragility was related to AD. Furthermore, a multitude of lipid-associated proteins exhibited statistical changes. In certain instances, the function of individual proteins affected the factors associated with them, whereas others demonstrated trends contrary to anticipated outcomes. These trends seem indicative of diverse feedback mechanisms that provide homeostatic equilibrium. The degree of unsaturation of fatty acids, correlated with cardiovascular risk, warrants specific attention. Certain bile acids exhibited the potential to cause vascular endothelial damage. Contemplating these discoveries in tandem with previously documented phenomena, subtle shifts in homeostatic functions seem to be linked to the fragility of vascular endothelial cells. This is evidenced by the slow and chronic evolution of Alzheimer's disease from preclinical stages to its manifestation.
Collapse
Affiliation(s)
- Suzumi M Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan
| | - Fumie Hamano
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan
- Life Sciences Core Facility, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Ayako Kobayashi
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan
| | - Shungo Adachi
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, 2-3-26 Aoumi, Koto-ku, Tokyo, 135-0064, Japan
| | - Tomohiro Andou
- Axcelead Drug Discovery Partners, Inc., 2-26-1 Muraoka-Higashi, Fujisawa, Kanagawa, 251-0012, Japan
| | - Tohru Natsume
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, 2-3-26 Aoumi, Koto-ku, Tokyo, 135-0064, Japan
| | - Yoshiya Oda
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan.
| |
Collapse
|
11
|
Dancy C, Heintzelman KE, Katt ME. The Glycocalyx: The Importance of Sugar Coating the Blood-Brain Barrier. Int J Mol Sci 2024; 25:8404. [PMID: 39125975 PMCID: PMC11312458 DOI: 10.3390/ijms25158404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The endothelial glycocalyx (GCX), located on the luminal surface of vascular endothelial cells, is composed of glycoproteins, proteoglycans, and glycosaminoglycans. It plays a pivotal role in maintaining blood-brain barrier (BBB) integrity and vascular health within the central nervous system (CNS), influencing critical processes such as blood flow regulation, inflammation modulation, and vascular permeability. While the GCX is ubiquitously expressed on the surface of every cell in the body, the GCX at the BBB is highly specialized, with a distinct composition of glycans, physical structure, and surface charge when compared to GCX elsewhere in the body. There is evidence that the GCX at the BBB is disrupted and partially shed in many diseases that impact the CNS. Despite this, the GCX has yet to be a major focus of therapeutic targeting for CNS diseases. This review examines diverse model systems used in cerebrovascular GCX-related research, emphasizing the importance of selecting appropriate models to ensure clinical relevance and translational potential. This review aims to highlight the importance of the GCX in disease and how targeting the GCX at the BBB specifically may be an effective approach for brain specific targeting for therapeutics.
Collapse
Affiliation(s)
- Candis Dancy
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
| | - Kaitlyn E. Heintzelman
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
12
|
Taghizadeh Ghassab F, Shamlou Mahmoudi F, Taheri Tinjani R, Emami Meibodi A, Zali MR, Yadegar A. Probiotics and the microbiota-gut-brain axis in neurodegeneration: Beneficial effects and mechanistic insights. Life Sci 2024; 350:122748. [PMID: 38843992 DOI: 10.1016/j.lfs.2024.122748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/21/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
Neurodegenerative diseases (NDs) are a group of heterogeneous disorders with a high socioeconomic burden. Although pharmacotherapy is currently the principal therapeutic approach for the management of NDs, mounting evidence supports the notion that the protracted application of available drugs would abate their dopaminergic outcomes in the long run. The therapeutic application of microbiome-based modalities has received escalating attention in biomedical works. In-depth investigations of the bidirectional communication between the microbiome in the gut and the brain offer a multitude of targets for the treatment of NDs or maximizing the patient's quality of life. Probiotic administration is a well-known microbial-oriented approach to modulate the gut microbiota and potentially influence the process of neurodegeneration. Of note, there is a strong need for further investigation to map out the mechanistic prospects for the gut-brain axis and the clinical efficacy of probiotics. In this review, we discuss the importance of microbiome modulation and hemostasis via probiotics, prebiotics, postbiotics and synbiotics in ameliorating pathological neurodegenerative events. Also, we meticulously describe the underlying mechanism of action of probiotics and their metabolites on the gut-brain axis in different NDs. We suppose that the present work will provide a functional direction for the use of probiotic-based modalities in promoting current practical treatments for the management of neurodegenerative-related diseases.
Collapse
Affiliation(s)
- Fatemeh Taghizadeh Ghassab
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shamlou Mahmoudi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Taheri Tinjani
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armitasadat Emami Meibodi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Rudroff T. Artificial Intelligence as a Replacement for Animal Experiments in Neurology: Potential, Progress, and Challenges. Neurol Int 2024; 16:805-820. [PMID: 39195562 DOI: 10.3390/neurolint16040060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Animal experimentation has long been a cornerstone of neurology research, but it faces growing scientific, ethical, and economic challenges. Advances in artificial intelligence (AI) are providing new opportunities to replace animal testing with more human-relevant and efficient methods. This article explores the potential of AI technologies such as brain organoids, computational models, and machine learning to revolutionize neurology research and reduce reliance on animal models. These approaches can better recapitulate human brain physiology, predict drug responses, and uncover novel insights into neurological disorders. They also offer faster, cheaper, and more ethical alternatives to animal experiments. Case studies demonstrate AI's ability to accelerate drug discovery for Alzheimer's, predict neurotoxicity, personalize treatments for Parkinson's, and restore movement in paralysis. While challenges remain in validating and integrating these technologies, the scientific, economic, practical, and moral advantages are driving a paradigm shift towards AI-based, animal-free research in neurology. With continued investment and collaboration across sectors, AI promises to accelerate the development of safer and more effective therapies for neurological conditions while significantly reducing animal use. The path forward requires the ongoing development and validation of these technologies, but a future in which they largely replace animal experiments in neurology appears increasingly likely. This transition heralds a new era of more humane, human-relevant, and innovative brain research.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
Mueller PM, Torres-Espín A, Vonder Haar C. Bayesian Methods: A Means of Improving Statistical Power in Preclinical Neurotrauma? Neurotrauma Rep 2024; 5:699-707. [PMID: 39071981 PMCID: PMC11271152 DOI: 10.1089/neur.2024.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
The field of neurotrauma is grappling with the effects of the recently identified replication crisis. As such, care must be taken to identify and perform the most appropriate statistical analyses. This will prevent misuse of research resources and ensure that conclusions are reasonable and within the scope of the data. We anticipate that Bayesian statistical methods will see increasing use in the coming years. Bayesian methods integrate prior beliefs (or prior data) into a statistical model to merge historical information and current experimental data. These methods may improve the ability to detect differences between experimental groups (i.e., statistical power) when used appropriately. However, researchers need to be aware of the strengths and limitations of such approaches if they are to implement or evaluate these analyses. Ultimately, an approach using Bayesian methodologies may have substantial benefits to statistical power, but caution needs to be taken when identifying and defining prior beliefs.
Collapse
Affiliation(s)
- Peyton M. Mueller
- Department of Neuroscience, Injury and Recovery Laboratory, Ohio State University, Columbus, Ohio, USA
| | - Abel Torres-Espín
- School of Public Health Sciences, University of Waterloo, Waterloo, Canada
| | - Cole Vonder Haar
- Department of Neuroscience, Injury and Recovery Laboratory, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
15
|
Pereira MF, Shyti R, Testa G. In and out: Benchmarking in vitro, in vivo, ex vivo, and xenografting approaches for an integrative brain disease modeling pipeline. Stem Cell Reports 2024; 19:767-795. [PMID: 38865969 PMCID: PMC11390705 DOI: 10.1016/j.stemcr.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 06/14/2024] Open
Abstract
Human cellular models and their neuronal derivatives have afforded unprecedented advances in elucidating pathogenic mechanisms of neuropsychiatric diseases. Notwithstanding their indispensable contribution, animal models remain the benchmark in neurobiological research. In an attempt to harness the best of both worlds, researchers have increasingly relied on human/animal chimeras by xenografting human cells into the animal brain. Despite the unparalleled potential of xenografting approaches in the study of the human brain, literature resources that systematically examine their significance and advantages are surprisingly lacking. We fill this gap by providing a comprehensive account of brain diseases that were thus far subjected to all three modeling approaches (transgenic rodents, in vitro human lineages, human-animal xenografting) and provide a critical appraisal of the impact of xenografting approaches for advancing our understanding of those diseases and brain development. Next, we give our perspective on integrating xenografting modeling pipeline with recent cutting-edge technological advancements.
Collapse
Affiliation(s)
- Marlene F Pereira
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| | - Reinald Shyti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Neurogenomics Centre, Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy.
| |
Collapse
|
16
|
Lacueva-Aparicio A, González VJ, Remacha AR, Woods D, Prado E, Ochoa I, Oliván S, Vázquez E. A microphysiological system for handling graphene related materials under flow conditions. NANOSCALE HORIZONS 2024; 9:990-1001. [PMID: 38606529 DOI: 10.1039/d4nh00064a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The field of nanotechnology has developed rapidly in recent decades due to its broad applications in many industrial and biomedical fields. Notably, 2D materials such as graphene-related materials (GRMs) have been extensively explored and, as such, their safety needs to be assessed. However, GRMs tend to deposit quickly, present low stability in aqueous solutions, and adsorb to plastic materials. Consequently, traditional approaches based on static assays facilitate their deposition and adsorption and fail to recreate human physiological conditions. Organ-on-a-chip (OOC) technology could, however, solve these drawbacks and lead to the development of microphysiological systems (MPSs) that mimic the microenvironment present in human tissues. In light of the above, in the present study a microfluidic system under flow conditions has been optimised to minimise graphene oxide (GO) and few-layer graphene (FLG) adsorption and deposition. For that purpose, a kidney-on-a-chip was developed and optimised to evaluate the effects of exposure to GO and FLG flakes at a sublethal dose under fluid flow conditions. In summary, MPSs are an innovative and precise tool for evaluating the effects of exposure to GRMs and other type of nanomaterials.
Collapse
Affiliation(s)
- Alodia Lacueva-Aparicio
- Tissue Microenvironment (TME) Lab, I3A _ IIS Aragón, University of Zaragoza, 50018 Zaragoza, Spain
- Instituto Regional de Investigación Científica Aplicada (IRICA), University of Castilla-La Mancha, Ciudad Real 13071, Spain.
| | - Viviana Jehová González
- Instituto Regional de Investigación Científica Aplicada (IRICA), University of Castilla-La Mancha, Ciudad Real 13071, Spain.
| | - Ana Rosa Remacha
- Tissue Microenvironment (TME) Lab, I3A _ IIS Aragón, University of Zaragoza, 50018 Zaragoza, Spain
| | - Daniel Woods
- Tissue Microenvironment (TME) Lab, I3A _ IIS Aragón, University of Zaragoza, 50018 Zaragoza, Spain
| | - Eduardo Prado
- Instituto Regional de Investigación Científica Aplicada (IRICA), University of Castilla-La Mancha, Ciudad Real 13071, Spain.
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, I3A _ IIS Aragón, University of Zaragoza, 50018 Zaragoza, Spain
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, I3A _ IIS Aragón, University of Zaragoza, 50018 Zaragoza, Spain
| | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA), University of Castilla-La Mancha, Ciudad Real 13071, Spain.
- Faculty of Chemical Science and Technology, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| |
Collapse
|
17
|
Tan LY, Cunliffe G, Hogan MP, Yeo XY, Oh C, Jin B, Kang J, Park J, Kwon MS, Kim M, Jung S. Emergence of the brain-border immune niches and their contribution to the development of neurodegenerative diseases. Front Immunol 2024; 15:1380063. [PMID: 38863704 PMCID: PMC11165048 DOI: 10.3389/fimmu.2024.1380063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Historically, the central nervous system (CNS) was regarded as 'immune-privileged', possessing its own distinct immune cell population. This immune privilege was thought to be established by a tight blood-brain barrier (BBB) and blood-cerebrospinal-fluid barrier (BCSFB), which prevented the crossing of peripheral immune cells and their secreted factors into the CNS parenchyma. However, recent studies have revealed the presence of peripheral immune cells in proximity to various brain-border niches such as the choroid plexus, cranial bone marrow (CBM), meninges, and perivascular spaces. Furthermore, emerging evidence suggests that peripheral immune cells may be able to infiltrate the brain through these sites and play significant roles in driving neuronal cell death and pathology progression in neurodegenerative disease. Thus, in this review, we explore how the brain-border immune niches may contribute to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We then discuss several emerging options for harnessing the neuroimmune potential of these niches to improve the prognosis and treatment of these debilitative disorders using novel insights from recent studies.
Collapse
Affiliation(s)
- Li Yang Tan
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Cunliffe
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael Patrick Hogan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chansik Oh
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Bohwan Jin
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junmo Kang
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junho Park
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Biomedical Science, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
18
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
19
|
Krut' VG, Kalinichenko AL, Maltsev DI, Jappy D, Shevchenko EK, Podgorny OV, Belousov VV. Optogenetic and chemogenetic approaches for modeling neurological disorders in vivo. Prog Neurobiol 2024; 235:102600. [PMID: 38548126 DOI: 10.1016/j.pneurobio.2024.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Animal models of human neurological disorders provide valuable experimental tools which enable us to study various aspects of disorder pathogeneses, ranging from structural abnormalities and disrupted metabolism and signaling to motor and mental deficits, and allow us to test novel therapies in preclinical studies. To be valid, these animal models should recapitulate complex pathological features at the molecular, cellular, tissue, and behavioral levels as closely as possible to those observed in human subjects. Pathological states resembling known human neurological disorders can be induced in animal species by toxins, genetic factors, lesioning, or exposure to extreme conditions. In recent years, novel animal models recapitulating neuropathologies in humans have been introduced. These animal models are based on synthetic biology approaches: opto- and chemogenetics. In this paper, we review recent opto- and chemogenetics-based animal models of human neurological disorders. These models allow for the creation of pathological states by disrupting specific processes at the cellular level. The artificial pathological states mimic a range of human neurological disorders, such as aging-related dementia, Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, and ataxias. Opto- and chemogenetics provide new opportunities unavailable with other animal models of human neurological disorders. These techniques enable researchers to induce neuropathological states varying in severity and ranging from acute to chronic. We also discuss future directions for the development and application of synthetic biology approaches for modeling neurological disorders.
Collapse
Affiliation(s)
- Viktoriya G Krut'
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Andrei L Kalinichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry I Maltsev
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Evgeny K Shevchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Oleg V Podgorny
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia.
| | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow 143025, Russia.
| |
Collapse
|
20
|
Mackay CA, Rath C, Rao S, Patole S. Plant-Derived Substances for Prevention of Necrotising Enterocolitis: A Systematic Review of Animal Studies. Nutrients 2024; 16:832. [PMID: 38542743 PMCID: PMC10975714 DOI: 10.3390/nu16060832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Inflammation, oxidative injury, and gut dysbiosis play an important role in the pathogenesis of necrotising enterocolitis (NEC). Plant-derived substances have historically been used as therapeutic agents due to their anti-inflammatory, antioxidant, and antimicrobial properties. We aimed to review pre-clinical evidence for plant-derived substances in the prevention and treatment of NEC. A systematic review was conducted using the following databases: PubMed, EMBASE, EMCARE, MEDLINE and Cochrane Library (PROSPERO CRD42022365477). Randomized controlled trials (RCTs) and quasi-RCTs that evaluated a plant-derived substance as an intervention for NEC in an animal model of the illness and compared pre-stated outcomes (e.g., clinical severity, severity of intestinal injury, mortality, laboratory markers of inflammation and oxidative injury) were included. Sixteen studies (n = 610) were included in the systematic review. Ten of the sixteen included RCTs (Preterm rat pups: 15, Mice: 1) reported mortality and all reported NEC-related histology. Meta-analysis showed decreased mortality [12/134 vs. 27/135; RR: 0.48 (95% CI: 0.26 to 0.87); p = 0.02, 10 RCTs] and decreased NEC in the experimental group [24/126 vs. 55/79; RR: 0.34 (95% CI: 0.22 to 0.52); p < 0.001, 6 RCTs]. Markers of inflammation (n = 11) and oxidative stress (n = 13) improved in all the studies that have reported this outcome. There was no significant publication bias for the outcome of mortality. Plant-derived substances have the potential to reduce the incidence and severity of histologically diagnosed NEC and mortality in rodent models. These findings are helpful in guiding further pre-clinical studies towards developing a food supplement for the prevention of NEC in preterm infants.
Collapse
Affiliation(s)
| | - Chandra Rath
- Neonatology, King Edward Memorial Hospita, Subiaco 6008, Australia
- Perth Children’s Hospital, Nedlands 6009, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| | - Shripada Rao
- Perth Children’s Hospital, Nedlands 6009, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| | - Sanjay Patole
- Neonatology, King Edward Memorial Hospita, Subiaco 6008, Australia
- School of Medicine, University of Western Australia, Crawley 6009, Australia
| |
Collapse
|
21
|
Shalash R, Levi-Ferber M, Cohen C, Dori A, Brodie C, Henis-Korenblit S. Cross-species modeling of muscular dystrophy in Caenorhabditis elegans using patient-derived extracellular vesicles. Dis Model Mech 2024; 17:dmm050412. [PMID: 38501170 PMCID: PMC11007864 DOI: 10.1242/dmm.050412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
Reliable disease models are critical for medicine advancement. Here, we established a versatile human disease model system using patient-derived extracellular vesicles (EVs), which transfer a pathology-inducing cargo from a patient to a recipient naïve model organism. As a proof of principle, we applied EVs from the serum of patients with muscular dystrophy to Caenorhabditis elegans and demonstrated their capability to induce a spectrum of muscle pathologies, including lifespan shortening and robust impairment of muscle organization and function. This demonstrates that patient-derived EVs can deliver disease-relevant pathologies between species and can be exploited for establishing novel and personalized models of human disease. Such models can potentially be used for disease diagnosis, prognosis, analyzing treatment responses, drug screening and identification of the disease-transmitting cargo of patient-derived EVs and their cellular targets. This system complements traditional genetic disease models and enables modeling of multifactorial diseases and of those not yet associated with specific genetic mutations.
Collapse
Affiliation(s)
- Rewayd Shalash
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Mor Levi-Ferber
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Coral Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
- The Mina and Everard Goodman Faculty of Life Sciences and Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Amir Dori
- Department of Neurology, Sheba Medical Center, Ramat-Gan 52621, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Chaya Brodie
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
- The Mina and Everard Goodman Faculty of Life Sciences and Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Sivan Henis-Korenblit
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
22
|
Rashidi AS, Tran DN, Peelen CR, van Gent M, Ouwendijk WJD, Verjans GMGM. Herpes simplex virus infection induces necroptosis of neurons and astrocytes in human fetal organotypic brain slice cultures. J Neuroinflammation 2024; 21:38. [PMID: 38302975 PMCID: PMC10832279 DOI: 10.1186/s12974-024-03027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Herpes simplex virus (HSV) encephalitis (HSE) is a serious and potentially life-threatening disease, affecting both adults and newborns. Progress in understanding the virus and host factors involved in neonatal HSE has been hampered by the limitations of current brain models that do not fully recapitulate the tissue structure and cell composition of the developing human brain in health and disease. Here, we developed a human fetal organotypic brain slice culture (hfOBSC) model and determined its value in mimicking the HSE neuropathology in vitro. METHODS Cell viability and tissues integrity were determined by lactate dehydrogenase release in supernatant and immunohistological (IHC) analyses. Brain slices were infected with green fluorescent protein (GFP-) expressing HSV-1 and HSV-2. Virus replication and spread were determined by confocal microscopy, PCR and virus culture. Expression of pro-inflammatory cytokines and chemokines were detected by PCR. Cell tropism and HSV-induced neuropathology were determined by IHC analysis. Finally, the in situ data of HSV-infected hfOBSC were compared to the neuropathology detected in human HSE brain sections. RESULTS Slicing and serum-free culture conditions were optimized to maintain the viability and tissue architecture of ex vivo human fetal brain slices for at least 14 days at 37 °C in a CO2 incubator. The hfOBSC supported productive HSV-1 and HSV-2 infection, involving predominantly infection of neurons and astrocytes, leading to expression of pro-inflammatory cytokines and chemokines. Both viruses induced programmed cell death-especially necroptosis-in infected brain slices at later time points after infection. The virus spread, cell tropism and role of programmed cell death in HSV-induced cell death resembled the neuropathology of HSE. CONCLUSIONS We developed a novel human brain culture model in which the viability of the major brain-resident cells-including neurons, microglia, astrocytes and oligodendrocytes-and the tissue architecture is maintained for at least 2 weeks in vitro under serum-free culture conditions. The close resemblance of cell tropism, spread and neurovirulence of HSV-1 and HSV-2 in the hfOBSC model with the neuropathological features of human HSE cases underscores its potential to detail the pathophysiology of other neurotropic viruses and as preclinical model to test novel therapeutic interventions.
Collapse
Affiliation(s)
- Ahmad S Rashidi
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Diana N Tran
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Caithlin R Peelen
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Michiel van Gent
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Werner J D Ouwendijk
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Georges M G M Verjans
- HerpesLabNL of the Department of Viroscience (Room Ee1720a), Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Sande R, Godad A, Doshi G. Zebrafish Experimental Animal Models for AD: A Comprehensive Review. Curr Rev Clin Exp Pharmacol 2024; 19:295-311. [PMID: 38284707 DOI: 10.2174/0127724328279684240104094257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 01/30/2024]
Abstract
AD disease (AD) is a multifaceted and intricate neurodegenerative disorder characterized by intracellular neurofibrillary tangle (NFT) formation and the excessive production and deposition of Aβ senile plaques. While transgenic AD models have been found instrumental in unravelling AD pathogenesis, they involve cost and time constraints during the preclinical phase. Zebrafish, owing to their simplicity, well-defined behavioural patterns, and relevance to neurodegenerative research, have emerged as a promising complementary model. Zebrafish possess glutaminergic and cholinergic pathways implicated in learning and memory, actively contributing to our understanding of neural transmission processes. This review sheds light on the molecular mechanisms by which various neurotoxic agents, including okadaic acid (OKA), cigarette smoke extract, metals, and transgenic zebrafish models with genetic similarities to AD patients, induce cognitive impairments and neuronal degeneration in mammalian systems. These insights may facilitate the identification of effective neurotoxic agents for replicating AD pathogenesis in the zebrafish brain. In this comprehensive review, the pivotal role of zebrafish models in advancing our comprehension of AD is emphasized. These models hold immense potential for shaping future research directions and clinical interventions, ultimately contributing to the development of novel AD therapies.
Collapse
Affiliation(s)
- Ruksar Sande
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Angel Godad
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| |
Collapse
|
24
|
Lisa DD, Muzzi L, Lagazzo A, Andolfi A, Martinoia S, Pastorino L. Long-term in vitroculture of 3D brain tissue model based on chitosan thermogel. Biofabrication 2023; 16:015011. [PMID: 37922538 DOI: 10.1088/1758-5090/ad0979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/03/2023] [Indexed: 11/07/2023]
Abstract
Methods for studying brain function and disease heavily rely onin vivoanimal models,ex-vivotissue slices, and 2D cell culture platforms. These methods all have limitations that significantly impact the clinical translatability of results. Consequently, models able to better recapitulate some aspects ofin vivohuman brain are needed as additional preclinical tools. In this context, 3D hydrogel-basedin vitromodels of the brain are considered promising tools. To create a 3D brain-on-a-chip model, a hydrogel capable of sustaining neuronal maturation over extended culture periods is required. Among biopolymeric hydrogels, chitosan-β-glycerophosphate (CHITO-β-GP) thermogels have demonstrated their versatility and applicability in the biomedical field over the years. In this study, we investigated the ability of this thermogel to encapsulate neuronal cells and support the functional maturation of a 3D neuronal network in long-term cultures. To the best of our knowledge, we demonstrated for the first time that CHITO-β-GP thermogel possesses optimal characteristics for promoting neuronal growth and the development of an electrophysiologically functional neuronal network derived from both primary rat neurons and neurons differentiated from human induced pluripotent stem cells (h-iPSCs) co-cultured with astrocytes. Specifically, two different formulations were firstly characterized by rheological, mechanical and injectability tests. Primary nervous cells and neurons differentiated from h-iPSCs were embedded into the two thermogel formulations. The 3D cultures were then deeply characterized by immunocytochemistry, confocal microscopy, and electrophysiological recordings, employing both 2D and 3D micro-electrode arrays. The thermogels supported the long-term culture of neuronal networks for up to 100 d. In conclusion, CHITO-β-GP thermogels exhibit excellent mechanical properties, stability over time under culture conditions, and bioactivity toward nervous cells. Therefore, they are excellent candidates as artificial extracellular matrices in brain-on-a-chip models, with applications in neurodegenerative disease modeling, drug screening, and neurotoxicity evaluation.
Collapse
Affiliation(s)
- Donatella Di Lisa
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Lorenzo Muzzi
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Alberto Lagazzo
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, via Montallegro 1, Genoa, Italy
| | - Andrea Andolfi
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Laura Pastorino
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| |
Collapse
|
25
|
Teli P, Kale V, Vaidya A. Beyond animal models: revolutionizing neurodegenerative disease modeling using 3D in vitro organoids, microfluidic chips, and bioprinting. Cell Tissue Res 2023; 394:75-91. [PMID: 37572163 DOI: 10.1007/s00441-023-03821-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by uncontrolled loss of neuronal cells leading to a progressive deterioration of brain functions. The transition rate of numerous neuroprotective drugs against Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, leading to FDA approval, is only 8-14% in the last two decades. Thus, in spite of encouraging preclinical results, these drugs have failed in human clinical trials, demonstrating that traditional cell cultures and animal models cannot accurately replicate human pathophysiology. Hence, in vitro three-dimensional (3D) models have been developed to bridge the gap between human and animal studies. Such technological advancements in 3D culture systems, such as human-induced pluripotent stem cell (iPSC)-derived cells/organoids, organ-on-a-chip technique, and 3D bioprinting, have aided our understanding of the pathophysiology and underlying mechanisms of human NDs. Despite these recent advances, we still lack a 3D model that recapitulates all the key aspects of NDs, thus making it difficult to study the ND's etiology in-depth. Hence in this review, we propose developing a combinatorial approach that allows the integration of patient-derived iPSCs/organoids with 3D bioprinting and organ-on-a-chip technique as it would encompass the neuronal cells along with their niche. Such a 3D combinatorial approach would characterize pathological processes thoroughly, making them better suited for high-throughput drug screening and developing effective novel therapeutics targeting NDs.
Collapse
Affiliation(s)
- Prajakta Teli
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India.
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India.
| |
Collapse
|
26
|
Gaspar L, Bartman S, Coppotelli G, Ross JM. Effect of apparatus characteristics on anxiety-like behavior in young adult and old mice of both sexes assessed by the elevated plus maze assay. Front Behav Neurosci 2023; 17:1182661. [PMID: 37638110 PMCID: PMC10450508 DOI: 10.3389/fnbeh.2023.1182661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Incidence of anxiety-like disorders in humans has been shown to decrease with aging; however, it is still under debate whether there are similarities in mice, which would support the use of mouse models in understanding the neuronal network changes that regulate anxiety-like behavior in aging. One of the most common tests used to assess anxiety-like behavior in laboratory animals is the elevated plus maze (EPM). Although several variables, such as room brightness and width of the maze arms, have been shown to influence the spontaneous animal behavior during the EPM test, none of these variables have ever been evaluated in aging to understand their possible differential effect on younger and older mice. We therefore decided to investigate the effect of apparatus construction on young adult and old mice of both sexes on EPM test performance. Our results show that distance traveled during the test is the variable that is most affected by apparatus characteristics independent of age and sex. We also found that apparatus construction was key in demonstrating that old mice spent more time and had relatively more entries in the open arms as compared to young mice, suggesting a decrease in anxiety-like behavior with age. Taken together, our data demonstrate that EPM apparatus characteristics dramatically affect test outcome with a wider arm apparatus being more effective in revealing age-dependent changes in anxiety-like behavior, thus, suggesting the use of a wider arm EPM when conducting aging studies in mice.
Collapse
Affiliation(s)
- Lauren Gaspar
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Sydney Bartman
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Giuseppe Coppotelli
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Jaime M. Ross
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
27
|
Ulaganathan S, Pitchaimani A. Spontaneous and familial models of Alzheimer's disease: Challenges and advances in preclinical research. Life Sci 2023:121918. [PMID: 37422070 DOI: 10.1016/j.lfs.2023.121918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder that is progressive and irreversible in nature. Even after decades of dedicated research and paradigm-shifting hypotheses of AD etiology, very few well-founded credible improvements have been foreseen in understanding the actual underlying mechanisms involved in the development of the disorder. As for any disease to be well-comprehended, AD also requires optimal modelling strategies, which will then pave way for effective therapeutic interventions. Most of the clinical trials and research towards better treatment of AD fail in translation, due to the inefficacy of explored animal models to mimic the actual AD pathology, precisely. The majority of the existing AD models are developed based on the mutations found in the familial form of AD (fAD) which accounts for less than 5 % of the incidence of AD. Further, the investigations also face more challenges due to the additional complexities and lacunae found in etiology of sporadic form of AD (sAD), which accounts for 95 % of total AD. This review illustrates the gaps found in different models of AD, both sporadic and familial variants with additional focus on recent avenues for accurate simulation of AD pathology using in vitro and chimeric AD models.
Collapse
Affiliation(s)
- Suryapriya Ulaganathan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, TN, India; School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, TN, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, TN, India; School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, TN, India.
| |
Collapse
|
28
|
Brown AR, Branthwaite HE, Farahbakhsh ZZ, Mukerjee S, Melugin PR, Song K, Noamany H, Siciliano CA. Structured tracking of alcohol reinforcement (STAR) for basic and translational alcohol research. Mol Psychiatry 2023; 28:1585-1598. [PMID: 36849824 PMCID: PMC10208967 DOI: 10.1038/s41380-023-01994-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
There is inherent tension between methodologies developed to address basic research questions in model species and those intended for preclinical to clinical translation: basic investigations require flexibility of experimental design as hypotheses are rapidly tested and revised, whereas preclinical models emphasize standardized protocols and specific outcome measures. This dichotomy is particularly relevant in alcohol research, which spans a diverse range of basic sciences in addition to intensive efforts towards understanding the pathophysiology of alcohol use disorder (AUD). To advance these goals there is a great need for approaches that facilitate synergy across basic and translational areas of nonhuman alcohol research. In male and female mice, we establish a modular alcohol reinforcement paradigm: Structured Tracking of Alcohol Reinforcement (STAR). STAR provides a robust platform for quantitative assessment of AUD-relevant behavioral domains within a flexible framework that allows direct crosstalk between translational and mechanistically oriented studies. To achieve cross-study integration, despite disparate task parameters, a straightforward multivariate phenotyping analysis is used to classify subjects based on propensity for heightened alcohol consumption and insensitivity to punishment. Combining STAR with extant preclinical alcohol models, we delineate longitudinal phenotype dynamics and reveal putative neuro-biomarkers of heightened alcohol use vulnerability via neurochemical profiling of cortical and brainstem tissues. Together, STAR allows quantification of time-resolved biobehavioral processes essential for basic research questions simultaneous with longitudinal phenotyping of clinically relevant outcomes, thereby providing a framework to facilitate cohesion and translation in alcohol research.
Collapse
Affiliation(s)
- Alex R Brown
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Hannah E Branthwaite
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zahra Z Farahbakhsh
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Snigdha Mukerjee
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Patrick R Melugin
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Keaton Song
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA
| | - Habiba Noamany
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
29
|
Alfalahi H, Dias SB, Khandoker AH, Chaudhuri KR, Hadjileontiadis LJ. A scoping review of neurodegenerative manifestations in explainable digital phenotyping. NPJ Parkinsons Dis 2023; 9:49. [PMID: 36997573 PMCID: PMC10063633 DOI: 10.1038/s41531-023-00494-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
Neurologists nowadays no longer view neurodegenerative diseases, like Parkinson's and Alzheimer's disease, as single entities, but rather as a spectrum of multifaceted symptoms with heterogeneous progression courses and treatment responses. The definition of the naturalistic behavioral repertoire of early neurodegenerative manifestations is still elusive, impeding early diagnosis and intervention. Central to this view is the role of artificial intelligence (AI) in reinforcing the depth of phenotypic information, thereby supporting the paradigm shift to precision medicine and personalized healthcare. This suggestion advocates the definition of disease subtypes in a new biomarker-supported nosology framework, yet without empirical consensus on standardization, reliability and interpretability. Although the well-defined neurodegenerative processes, linked to a triad of motor and non-motor preclinical symptoms, are detected by clinical intuition, we undertake an unbiased data-driven approach to identify different patterns of neuropathology distribution based on the naturalistic behavior data inherent to populations in-the-wild. We appraise the role of remote technologies in the definition of digital phenotyping specific to brain-, body- and social-level neurodegenerative subtle symptoms, emphasizing inter- and intra-patient variability powered by deep learning. As such, the present review endeavors to exploit digital technologies and AI to create disease-specific phenotypic explanations, facilitating the understanding of neurodegenerative diseases as "bio-psycho-social" conditions. Not only does this translational effort within explainable digital phenotyping foster the understanding of disease-induced traits, but it also enhances diagnostic and, eventually, treatment personalization.
Collapse
Affiliation(s)
- Hessa Alfalahi
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| | - Sofia B Dias
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- CIPER, Faculdade de Motricidade Humana, University of Lisbon, Lisbon, Portugal
| | - Ahsan H Khandoker
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Kallol Ray Chaudhuri
- Parkinson Foundation, International Center of Excellence, King's College London, Denmark Hills, London, UK
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Leontios J Hadjileontiadis
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Electrical and Computer Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
30
|
Lesicka M, Dmitrzak-Weglarz M, Jablonska E, Wieczorek E, Kapelski P, Szczepankiewicz A, Pawlak J, Reszka E. Methylation of melatonin receptors in patients with unipolar and bipolar depression. Mech Ageing Dev 2023; 211:111776. [PMID: 36706965 DOI: 10.1016/j.mad.2023.111776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 01/25/2023]
Abstract
Disturbances of melatonin secretion alter the circadian rhythm and sleep-wake cycle, which is observed among patients with depression. Melatonin acts via melatonin receptors MT1 and MT2, which are present in many tissues, including peripheral blood mononuclear cells (PBMC). We assume that disturbances of the melatonin pathway in the brain may be reflected by molecular changes in peripheral organs. The study objective was to evaluate the methylation profile of CpG island in the promoter region of melatonin receptor genes MTNR1A and MTNR1B in PBMC of patients with depression and compare it with healthy volunteers. The study group comprised 85 patients with unipolar (UP) and bipolar disorders (BP) and 83 controls. The methylation pattern of CpG island in the promoter region was analyzed using the quantitative methylation-specific real-time PCR (qMSP-PCR) method. We found that the methylation profile of the patients with depression varied in comparison to the control group. The methylation level of MTNR1A was significantly lower among depressed patients compared to controls. Additionally, melatonin concentration was negatively correlated with MTNR1B methylation level among the UP patients. The study may suggest that the methylation profile of melatonin receptors in PBMC may be used as a complementary molecular marker in depression diagnosis.
Collapse
Affiliation(s)
- Monika Lesicka
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland.
| | - Monika Dmitrzak-Weglarz
- Department of Psychiatric Genetics, Department of Psychiatry, University of Medical Sciences, Poznan, Poland
| | - Ewa Jablonska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Edyta Wieczorek
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Pawel Kapelski
- Department of Psychiatric Genetics, Department of Psychiatry, University of Medical Sciences, Poznan, Poland
| | - Aleksandra Szczepankiewicz
- Department of Psychiatric Genetics, Department of Psychiatry, University of Medical Sciences, Poznan, Poland
| | - Joanna Pawlak
- Department of Psychiatric Genetics, Department of Psychiatry, University of Medical Sciences, Poznan, Poland
| | - Edyta Reszka
- Department of Translational Research, Nofer Institute of Occupational Medicine, Lodz, Poland
| |
Collapse
|
31
|
In Vitro 3D Modeling of Neurodegenerative Diseases. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010093. [PMID: 36671665 PMCID: PMC9855033 DOI: 10.3390/bioengineering10010093] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
The study of neurodegenerative diseases (such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis) is very complex due to the difficulty in investigating the cellular dynamics within nervous tissue. Despite numerous advances in the in vivo study of these diseases, the use of in vitro analyses is proving to be a valuable tool to better understand the mechanisms implicated in these diseases. Although neural cells remain difficult to obtain from patient tissues, access to induced multipotent stem cell production now makes it possible to generate virtually all neural cells involved in these diseases (from neurons to glial cells). Many original 3D culture model approaches are currently being developed (using these different cell types together) to closely mimic degenerative nervous tissue environments. The aim of these approaches is to allow an interaction between glial cells and neurons, which reproduces pathophysiological reality by co-culturing them in structures that recapitulate embryonic development or facilitate axonal migration, local molecule exchange, and myelination (to name a few). This review details the advantages and disadvantages of techniques using scaffolds, spheroids, organoids, 3D bioprinting, microfluidic systems, and organ-on-a-chip strategies to model neurodegenerative diseases.
Collapse
|
32
|
Microglial Expression of the Wnt Signaling Modulator DKK2 Differs between Human Alzheimer's Disease Brains and Mouse Neurodegeneration Models. eNeuro 2023; 10:ENEURO.0306-22.2022. [PMID: 36599670 PMCID: PMC9836029 DOI: 10.1523/eneuro.0306-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023] Open
Abstract
Wnt signaling is crucial for synapse and cognitive function. Indeed, deficient Wnt signaling is causally related to increased expression of DKK1, an endogenous negative Wnt regulator, and synapse loss, both of which likely contribute to cognitive decline in Alzheimer's disease (AD). Increasingly, AD research efforts have probed the neuroinflammatory role of microglia, the resident immune cells of the CNS, which have furthermore been shown to be modulated by Wnt signaling. The DKK1 homolog DKK2 has been previously identified as an activated response and/or disease-associated microglia (DAM/ARM) gene in a mouse model of AD. Here, we performed a detailed analysis of DKK2 in mouse models of neurodegeneration, and in human AD brain. In APP/PS1 and APPNL-G-F AD mouse model brains as well as in SOD1G93A ALS mouse model spinal cords, but not in control littermates, we demonstrated significant microgliosis and microglial Dkk2 mRNA upregulation in a disease-stage-dependent manner. In the AD models, these DAM/ARM Dkk2+ microglia preferentially accumulated close to βAmyloid plaques. Furthermore, recombinant DKK2 treatment of rat hippocampal primary neurons blocked WNT7a-induced dendritic spine and synapse formation, indicative of an anti-synaptic effect similar to that of DKK1. In stark contrast, no such microglial DKK2 upregulation was detected in the postmortem human frontal cortex from individuals diagnosed with AD or pathologic aging. In summary, the difference in microglial expression of the DAM/ARM gene DKK2 between mouse models and human AD brain highlights the increasingly recognized limitations of using mouse models to recapitulate facets of human neurodegenerative disease.
Collapse
|
33
|
Xu J, Zheng Y, Wang L, Liu Y, Wang X, Li Y, Chi G. miR-124: A Promising Therapeutic Target for Central Nervous System Injuries and Diseases. Cell Mol Neurobiol 2022; 42:2031-2053. [PMID: 33886036 DOI: 10.1007/s10571-021-01091-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Central nervous system injuries and diseases, such as ischemic stroke, spinal cord injury, neurodegenerative diseases, glioblastoma, multiple sclerosis, and the resulting neuroinflammation often lead to death or long-term disability. MicroRNAs are small, non-coding, single-stranded RNAs that regulate posttranscriptional gene expression in both physiological and pathological cellular processes, including central nervous system injuries and disorders. Studies on miR-124, one of the most abundant microRNAs in the central nervous system, have shown that its dysregulation is related to the occurrence and development of pathology within the central nervous system. Herein, we review the molecular regulatory functions, underlying mechanisms, and effective delivery methods of miR-124 in the central nervous system, where it is involved in pathological conditions. The review also provides novel insights into the therapeutic target potential of miR-124 in the treatment of human central nervous system injuries or diseases.
Collapse
Affiliation(s)
- Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Yangyang Zheng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China
| | - Liangjia Wang
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yining Liu
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Xishu Wang
- Clinical Medical College, Jilin University, Changchun, 130000, People's Republic of China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130000, People's Republic of China.
| |
Collapse
|
34
|
Buckles VD, Xiong C, Bateman RJ, Hassenstab J, Allegri R, Berman SB, Chhatwal JP, Danek A, Fagan AM, Ghetti B, Goate A, Graff-Radford N, Jucker M, Levin J, Marcus DS, Masters CL, McCue L, McDade E, Mori H, Moulder KL, Noble JM, Paumier K, Preische O, Ringman JM, Fox NC, Salloway S, Schofield PR, Martins R, Vöglein J, Morris JC. Different rates of cognitive decline in autosomal dominant and late-onset Alzheimer disease. Alzheimers Dement 2022; 18:1754-1764. [PMID: 34854530 PMCID: PMC9160203 DOI: 10.1002/alz.12505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/13/2021] [Accepted: 09/22/2021] [Indexed: 01/28/2023]
Abstract
As prevention trials advance with autosomal dominant Alzheimer disease (ADAD) participants, understanding the similarities and differences between ADAD and "sporadic" late-onset AD (LOAD) is critical to determine generalizability of findings between these cohorts. Cognitive trajectories of ADAD mutation carriers (MCs) and autopsy-confirmed LOAD individuals were compared to address this question. Longitudinal rates of change on cognitive measures were compared in ADAD MCs (n = 310) and autopsy-confirmed LOAD participants (n = 163) before and after symptom onset (estimated/observed). LOAD participants declined more rapidly in the presymptomatic (preclinical) period and performed more poorly at symptom onset than ADAD participants on a cognitive composite. After symptom onset, however, the younger ADAD MCs declined more rapidly. The similar but not identical cognitive trajectories (declining but at different rates) for ADAD and LOAD suggest common AD pathologies but with some differences.
Collapse
Affiliation(s)
- Virginia D. Buckles
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ricardo Allegri
- Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Sarah B. Berman
- Department of Neurology and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jasmeer P. Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Klinikum der Universität München, Munich Germany
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Mathias Jucker
- DZNE Tuebingen & Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Johannes Levin
- DZNE Munich, Munich Cluster of systems neurology (SyNergy) & Ludwig-Maximilians-Universität, Munich, Germany
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Lena McCue
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hiroshi Mori
- Department of Neuroscience, Osaka City University Medical School, Osaka City, Japan
| | - Krista L. Moulder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Aging Brain, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Katrina Paumier
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Oliver Preische
- DZNE Tuebingen & University of Tuebingen, Tuebingen, Germany
| | - John M. Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nick C Fox
- Department of Neurodegenerative Disease & UK Dementia Research Institute, Institute of Neurology, London, UK
| | - Stephen Salloway
- Department of Neurology, Butler Hospital & Alpert Medical School of Brown University, Providence, RI, 02906, USA
| | - Peter R. Schofield
- Neuroscience Research Australia & School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Ralph Martins
- Sir James McCusker Alzheimer’s Disease Research Unit, Edith Cowan University, Nedlands, Australia
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE) and Department of Neurology, Ludwig-Maximilians Universität München; Munich, Germany
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | |
Collapse
|
35
|
iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. J Pers Med 2022; 12:jpm12091485. [PMID: 36143270 PMCID: PMC9500601 DOI: 10.3390/jpm12091485] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022] Open
Abstract
In the past, several animal disease models were developed to study the molecular mechanism of neurological diseases and discover new therapies, but the lack of equivalent animal models has minimized the success rate. A number of critical issues remain unresolved, such as high costs for developing animal models, ethical issues, and lack of resemblance with human disease. Due to poor initial screening and assessment of the molecules, more than 90% of drugs fail during the final step of the human clinical trial. To overcome these limitations, a new approach has been developed based on induced pluripotent stem cells (iPSCs). The discovery of iPSCs has provided a new roadmap for clinical translation research and regeneration therapy. In this article, we discuss the potential role of patient-derived iPSCs in neurological diseases and their contribution to scientific and clinical research for developing disease models and for developing a roadmap for future medicine. The contribution of humaniPSCs in the most common neurodegenerative diseases (e.g., Parkinson’s disease and Alzheimer’s disease, diabetic neuropathy, stroke, and spinal cord injury) were examined and ranked as per their published literature on PUBMED. We have observed that Parkinson’s disease scored highest, followed by Alzheimer’s disease. Furthermore, we also explored recent advancements in the field of personalized medicine, such as the patient-on-a-chip concept, where iPSCs can be grown on 3D matrices inside microfluidic devices to create an in vitro disease model for personalized medicine.
Collapse
|
36
|
Dias A, Silva L, Moura J, Gabriel D, Maia LF. Fluid biomarkers in stroke: From animal models to clinical care. Acta Neurol Scand 2022; 146:332-347. [PMID: 35838031 DOI: 10.1111/ane.13668] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022]
Abstract
Stroke is a leading cause of death and disability worldwide. Stroke prevention, early diagnosis, and efficient acute treatment are priorities to successfully impact stroke death and disability. Fluid biomarkers may improve stroke differential diagnostic, patient stratification for acute treatment, and post-stroke individualized rehabilitation. In the present work, we characterized the use of stroke animal models in fluid biomarker research through a systematic review of PubMed and Scopus databases, followed by a literature review on the translation to the human stroke care setting and future perspectives in the field. We found increasing numbers of publications but with limited translation to the clinic. Animal studies are very heterogeneous, do not account for several human features present in stroke, and, importantly, only a minority of such studies used human cohorts to validate biomarker findings. Clinical studies have found appealing candidates, both protein and circulating nucleic acids, to contribute to a more personalized stroke care pathway. Still, brain tissue complexity and the fact that different brain pathologies share lesion biomarkers make this task challenging due to biomarker low specificity. Moreover, the study design and lack of validation cohorts may have precluded a formal integration of biomarkers in different steps of stroke diagnosis and treatment. To overcome such issues, recent pivotal studies on biomarker dynamics in individual patients are providing added value to diagnosis and anticipating patients' early prognosis. Presently, the most consistent protein biomarkers for stroke diagnosis and short- and long-term prognosis are associated with tissue damage at neuronal (TAU), axonal (NFL), or astroglial (GFAP and S100β) levels. Most promising nucleic acids are microRNAs (miR), due to their stability in plasma and ease of access. Still, clinical validation and standardized quantitation place them a step behind compared protein as stroke biomarkers. Ultimately, the definition of clinically relevant biomarker panels and optimization of fast and sensitive biomarker measurements in the blood, together with their combination with clinical and neuroimaging data, will pave the way toward personalized stroke care.
Collapse
Affiliation(s)
- Alexandre Dias
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Lénia Silva
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - João Moura
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Denis Gabriel
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Luis F Maia
- Department of Neurology, Centro Hospitalar Universitário do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| |
Collapse
|
37
|
Legault EM, Bouquety J, Drouin-Ouellet J. Disease Modeling of Neurodegenerative Disorders Using Direct Neural Reprogramming. Cell Reprogram 2022; 24:228-251. [PMID: 35749150 DOI: 10.1089/cell.2021.0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Understanding the pathophysiology of CNS-associated neurological diseases has been hampered by the inaccessibility of patient brain tissue to perform live analyses at the molecular level. To this end, neural cells obtained by differentiation of patient-derived induced pluripotent stem cells (iPSCs) are considerably helpful, especially in the context of monogenic-based disorders. More recently, the use of direct reprogramming to convert somatic cells to neural cells has emerged as an alternative to iPSCs to generate neurons, astrocytes, and oligodendrocytes. This review focuses on the different studies that used direct neural reprogramming to study disease-associated phenotypes in the context of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
| | - Julie Bouquety
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | |
Collapse
|
38
|
Abstract
SignificanceSingle-cell transcriptomics has revealed specific glial activation states associated with the pathogenesis of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease (AD and PD). What is still needed are clinically relevant biomarkers for deciphering such glial states in AD and PD patients. To this end, we applied proteome analysis in cerebrospinal fluid (CSF) of mouse models of AD and PD pathology. This allowed us to identify a panel of glial CSF proteins that largely match the transcriptomic changes. The identified proteins can also be quantified in human CSF and show changes in AD patients, supporting their relevance as biomarker candidates to stage glial activation in patients with neurodegenerative diseases.
Collapse
|
39
|
Teli P, Kale V, Vaidya A. Mesenchymal stromal cells-derived secretome protects Neuro-2a cells from oxidative stress-induced loss of neurogenesis. Exp Neurol 2022; 354:114107. [PMID: 35551901 DOI: 10.1016/j.expneurol.2022.114107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/20/2022] [Accepted: 05/02/2022] [Indexed: 12/01/2022]
Abstract
Neurodegenerative diseases (ND) are characterized by debilitating medical conditions that principally affect the neuronal cells in the human brain. One of the major reasons that there are no effective drugs for the treatment of ND is because researchers face technical challenges while conducting studies to understand the molecular mechanism behind ND. Although various studies have established in vitro neurodegenerative model systems, we feel that these model systems are not physiologically relevant, as they do not mimic the in vivo situation of chronic insult. Therefore, the primary aim of this study was to establish an in vitro neurodegenerative model system by inducing oxidative stress in such a way that the neuronal cells remain viable, but lose their structural and functional characteristics. Using a murine neuroblastoma cell line, Neuro-2a, we demonstrate that induction of oxidative stress significantly affects various neurite outgrowth parameters and reduces the expression of neuronal and autophagy markers without causing apoptosis in them. Previously, we have discussed the possible therapeutic applications of mesenchymal stromal cells (MSCs) and their secretome in the treatment of ND. Here, using two distinct approaches, we show that when Neuro-2a cells subjected to oxidative stress are exposed to MSC-derived conditioned medium (secretome), they exhibit a significant improvement in various neuronal parameters and in the expression of neuronal markers. Overall, our findings support the salutary role of MSC-derived secretome in rescuing the oxidative stress-induced loss of neurogenesis using a physiologically relevant in vitro model system. Our data underscore the propensity of the MSC-secretome in reversing ND.
Collapse
Affiliation(s)
- Prajakta Teli
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India; Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India; Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune 412115, India; Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune 412115, India.
| |
Collapse
|
40
|
Inducible Animal Models of Skin Fibrosis; Updated Review of the Literature. JORJANI BIOMEDICINE JOURNAL 2022. [DOI: 10.52547/jorjanibiomedj.10.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
41
|
Tello JA, Williams HE, Eppler RM, Steinhilb ML, Khanna M. Animal Models of Neurodegenerative Disease: Recent Advances in Fly Highlight Innovative Approaches to Drug Discovery. Front Mol Neurosci 2022; 15:883358. [PMID: 35514431 PMCID: PMC9063566 DOI: 10.3389/fnmol.2022.883358] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases represent a formidable challenge to global health. As advances in other areas of medicine grant healthy living into later decades of life, aging diseases such as Alzheimer's disease (AD) and other neurodegenerative disorders can diminish the quality of these additional years, owed largely to the lack of efficacious treatments and the absence of durable cures. Alzheimer's disease prevalence is predicted to more than double in the next 30 years, affecting nearly 15 million Americans, with AD-associated costs exceeding $1 billion by 2050. Delaying onset of AD and other neurodegenerative diseases is critical to improving the quality of life for patients and reducing the burden of disease on caregivers and healthcare systems. Significant progress has been made to model disease pathogenesis and identify points of therapeutic intervention. While some researchers have contributed to our understanding of the proteins and pathways that drive biological dysfunction in disease using in vitro and in vivo models, others have provided mathematical, biophysical, and computational technologies to identify potential therapeutic compounds using in silico modeling. The most exciting phase of the drug discovery process is now: by applying a target-directed approach that leverages the strengths of multiple techniques and validates lead hits using Drosophila as an animal model of disease, we are on the fast-track to identifying novel therapeutics to restore health to those impacted by neurodegenerative disease.
Collapse
Affiliation(s)
- Judith A. Tello
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Haley E. Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
| | - Robert M. Eppler
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - Michelle L. Steinhilb
- Department of Biology, Central Michigan University, Mount Pleasant, MI, United States
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center of Innovation in Brain Science, Tucson, AZ, United States
- Department of Molecular Pathobiology, New York University, New York, NY, United States
| |
Collapse
|
42
|
Doyle JM, Croll RP. A Critical Review of Zebrafish Models of Parkinson's Disease. Front Pharmacol 2022; 13:835827. [PMID: 35370740 PMCID: PMC8965100 DOI: 10.3389/fphar.2022.835827] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
A wide variety of human diseases have been modelled in zebrafish, including various types of cancer, cardiovascular diseases and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Recent reviews have summarized the currently available zebrafish models of Parkinson’s Disease, which include gene-based, chemically induced and chemogenetic ablation models. The present review updates the literature, critically evaluates each of the available models of Parkinson’s Disease in zebrafish and compares them with similar models in invertebrates and mammals to determine their advantages and disadvantages. We examine gene-based models, including ones linked to Early-Onset Parkinson’s Disease: PARKIN, PINK1, DJ-1, and SNCA; but we also examine LRRK2, which is linked to Late-Onset Parkinson’s Disease. We evaluate chemically induced models like MPTP, 6-OHDA, rotenone and paraquat, as well as chemogenetic ablation models like metronidazole-nitroreductase. The article also reviews the unique advantages of zebrafish, including the abundance of behavioural assays available to researchers and the efficiency of high-throughput screens. This offers a rare opportunity for assessing the potential therapeutic efficacy of pharmacological interventions. Zebrafish also are very amenable to genetic manipulation using a wide variety of techniques, which can be combined with an array of advanced microscopic imaging methods to enable in vivo visualization of cells and tissue. Taken together, these factors place zebrafish on the forefront of research as a versatile model for investigating disease states. The end goal of this review is to determine the benefits of using zebrafish in comparison to utilising other animals and to consider the limitations of zebrafish for investigating human disease.
Collapse
Affiliation(s)
- Jillian M Doyle
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
43
|
Prokop JW, Jdanov V, Savage L, Morris M, Lamb N, VanSickle E, Stenger CL, Rajasekaran S, Bupp CP. Computational and Experimental Analysis of Genetic Variants. Compr Physiol 2022; 12:3303-3336. [PMID: 35578967 DOI: 10.1002/cphy.c210012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genomics has grown exponentially over the last decade. Common variants are associated with physiological changes through statistical strategies such as Genome-Wide Association Studies (GWAS) and quantitative trail loci (QTL). Rare variants are associated with diseases through extensive filtering tools, including population genomics and trio-based sequencing (parents and probands). However, the genomic associations require follow-up analyses to narrow causal variants, identify genes that are influenced, and to determine the physiological changes. Large quantities of data exist that can be used to connect variants to gene changes, cell types, protein pathways, clinical phenotypes, and animal models that establish physiological genomics. This data combined with bioinformatics including evolutionary analysis, structural insights, and gene regulation can yield testable hypotheses for mechanisms of genomic variants. Molecular biology, biochemistry, cell culture, CRISPR editing, and animal models can test the hypotheses to give molecular variant mechanisms. Variant characterizations can be a significant component of educating future professionals at the undergraduate, graduate, or medical training programs through teaching the basic concepts and terminology of genetics while learning independent research hypothesis design. This article goes through the computational and experimental analysis strategies of variant characterization and provides examples of these tools applied in publications. © 2022 American Physiological Society. Compr Physiol 12:3303-3336, 2022.
Collapse
Affiliation(s)
- Jeremy W Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Vladislav Jdanov
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Lane Savage
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Michele Morris
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Neil Lamb
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | | | - Cynthia L Stenger
- Department of Mathematics, University of North Alabama, Florence, Alabama, USA
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Pediatric Intensive Care Unit, Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA.,Office of Research, Spectrum Health, Grand Rapids, Michigan, USA
| | - Caleb P Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Medical Genetics, Spectrum Health, Grand Rapids, Michigan, USA
| |
Collapse
|
44
|
Tian R, Han K, Geng Y, Yang C, Shi C, Thomas PB, Pearce C, Moffatt K, Ma S, Xu S, Yang G, Zhou X, Gladyshev VN, Liu X, Fisher DO, Chopin LK, Leiner NO, Baker AM, Fan G, Seim I. A chromosome-level genome of Antechinus flavipes provides a reference for an Australian marsupial genus with male death after mating. Mol Ecol Resour 2022; 22:740-754. [PMID: 34486812 PMCID: PMC9290055 DOI: 10.1111/1755-0998.13501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/16/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022]
Abstract
The 15 species of small carnivorous marsupials that comprise the genus Antechinus exhibit semelparity, a rare life-history strategy in mammals where synchronized death occurs after one breeding season. Antechinus males, but not females, age rapidly (demonstrate organismal senescence) during the breeding season and show promise as new animal models of ageing. Some antechinus species are also threatened or endangered. Here, we report a chromosome-level genome of a male yellow-footed antechinus Antechinus flavipes. The genome assembly has a total length of 3.2 Gb with a contig N50 of 51.8 Mb and a scaffold N50 of 636.7 Mb. We anchored and oriented 99.7% of the assembly on seven pseudochromosomes and found that repetitive DNA sequences occupy 51.8% of the genome. Draft genome assemblies of three related species in the subfamily Phascogalinae, two additional antechinus species (Antechinus argentus and A. arktos) and the iteroparous sister species Murexia melanurus, were also generated. Preliminary demographic analysis supports the hypothesis that climate change during the Pleistocene isolated species in Phascogalinae and shaped their population size. A transcriptomic profile across the A. flavipes breeding season allowed us to identify genes associated with aspects of the male die-off. The chromosome-level A. flavipes genome provides a steppingstone to understanding an enigmatic life-history strategy and a resource to assist the conservation of antechinuses.
Collapse
Affiliation(s)
- Ran Tian
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Kai Han
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
| | - Yuepan Geng
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Chen Yang
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | | | - Patrick B. Thomas
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- Queensland Bladder Cancer InitiativeTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyWoolloongabbaQLDAustralia
| | - Coral Pearce
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| | - Kate Moffatt
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| | - Siming Ma
- Genome Institute of SingaporeAgency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Guang Yang
- Jiangsu Key Laboratory for Biodiversity and BiotechnologyCollege of Life SciencesNanjing Normal UniversityNanjingChina
| | - Xuming Zhou
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMAUSA
| | - Xin Liu
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
| | - Diana O. Fisher
- School of Biological SciencesUniversity of QueenslandBrisbaneQLDAustralia
| | - Lisa K. Chopin
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
| | - Natália O. Leiner
- Laboratório de Ecologia de MamíferosInstituto de BiologiaUniversidade Federal de UberlândiaUberlândiaBrazil
| | - Andrew M. Baker
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
- Natural Environments ProgramQueensland MuseumSouth BrisbaneQLDAustralia
| | - Guangyi Fan
- BGI‐QingdaoBGI‐ShenzhenQingdaoChina
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacauChina
- State Key Laboratory of Agricultural GenomicsBGI‐ShenzhenShenzhenChina
| | - Inge Seim
- Integrative Biology LaboratoryCollege of Life SciencesNanjing Normal UniversityNanjingChina
- Ghrelin Research GroupTranslational Research Institute‐Institute of Health and Biomedical InnovationSchool of Biomedical SciencesQueensland University of TechnologyBrisbaneQLDAustralia
- Australian Prostate Cancer Research Centre‐QueenslandTranslational Research Institute – Institute of Health and Biomedical InnovationQueensland University of TechnologyBrisbaneQLDAustralia
- School of Biology and Environmental ScienceQueensland University of TechnologyBrisbaneQLDAustralia
| |
Collapse
|
45
|
Moore MJ, Demeyere N. Lesion symptom mapping of domain-specific cognitive impairments using routine imaging in stroke. Neuropsychologia 2022; 167:108159. [PMID: 35041840 DOI: 10.1016/j.neuropsychologia.2022.108159] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/14/2021] [Accepted: 01/12/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION This large-scale lesion-symptom mapping study investigates the necessary neuro-anatomical substrates of 5 cognitive domains frequently affected post stroke: Language, Attention, Praxis, Number, and Memory. This study aims to demonstrate the validity of using routine clinical brain imaging and standard bedside cognitive screening data from a large, real-world patient cohort for lesion-symptom mapping. PATIENTS AND METHODS Behavioural cognitive screening data from the Oxford Cognitive Screen and routine clinical neuroimaging from 573 acute patients was used in voxel-based lesion-symptom mapping analyses. Patients were classed as impaired or not on each of the subtests within 5 cognitive domains. RESULTS Distinct patterns of lesion damage were associated with different domains. Language functions were associated with damage to left hemisphere fronto-temporal areas. Visuo-spatial functions were associated with damage to posterior occipital areas (Visual Field) and the right temporo-parietal region (Visual Neglect). Different memory impairments were linked to distinct voxel clusters within the left insular and opercular cortices. Deficits which were not associated with localised voxels (e.g. executive function, praxis) represent distributed, bilateral functions. DISCUSSION The standardised, brief Oxford Cognitive Screen was able to reliably differentiate distinct neural correlates critically involved in supporting domain-specific cognitive abilities. CONCLUSION By demonstrating and replicating known brain anatomy correlates within real-life clinical cohorts using routinely collected scans and standard bedside screens, we open up VLSM techniques to a wealth of clinically relevant studies which can capitalise on using existing clinical data.
Collapse
Affiliation(s)
- Margaret Jane Moore
- University of Oxford, Department of Experimental Psychology, Radcliffe Observatory Quarter, Oxford, OX2 6GG, United Kingdom
| | - Nele Demeyere
- University of Oxford, Department of Experimental Psychology, Radcliffe Observatory Quarter, Oxford, OX2 6GG, United Kingdom.
| |
Collapse
|
46
|
Ozgun A, Lomboni D, Arnott H, Staines WA, Woulfe J, Variola F. Biomaterial-based strategies for in vitro neural models. Biomater Sci 2022; 10:1134-1165. [PMID: 35023513 DOI: 10.1039/d1bm01361k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In vitro models have been used as a complementary tool to animal studies in understanding the nervous system's physiological mechanisms and pathological disorders, while also serving as platforms to evaluate the safety and efficiency of therapeutic candidates. Following recent advances in materials science, micro- and nanofabrication techniques and cell culture systems, in vitro technologies have been rapidly gaining the potential to bridge the gap between animal and clinical studies by providing more sophisticated models that recapitulate key aspects of the structure, biochemistry, biomechanics, and functions of human tissues. This was made possible, in large part, by the development of biomaterials that provide cells with physicochemical features that closely mimic the cellular microenvironment of native tissues. Due to the well-known material-driven cellular response and the importance of mimicking the environment of the target tissue, the selection of optimal biomaterials represents an important early step in the design of biomimetic systems to investigate brain structures and functions. This review provides a comprehensive compendium of commonly used biomaterials as well as the different fabrication techniques employed for the design of neural tissue models. Furthermore, the authors discuss the main parameters that need to be considered to develop functional platforms not only for the study of brain physiological functions and pathological processes but also for drug discovery/development and the optimization of biomaterials for neural tissue engineering.
Collapse
Affiliation(s)
- Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - David Lomboni
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Hallie Arnott
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - William A Staines
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - John Woulfe
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada. .,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada.,The Ottawa Hospital, Ottawa, Canada.,Children's Hospital of Eastern Ontario (CHEO), Ottawa, Canada
| |
Collapse
|
47
|
Fanizza F, Campanile M, Forloni G, Giordano C, Albani D. Induced pluripotent stem cell-based organ-on-a-chip as personalized drug screening tools: A focus on neurodegenerative disorders. J Tissue Eng 2022; 13:20417314221095339. [PMID: 35570845 PMCID: PMC9092580 DOI: 10.1177/20417314221095339] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/04/2022] [Indexed: 01/15/2023] Open
Abstract
The Organ-on-a-Chip (OoC) technology shows great potential to revolutionize the drugs development pipeline by mimicking the physiological environment and functions of human organs. The translational value of OoC is further enhanced when combined with patient-specific induced pluripotent stem cells (iPSCs) to develop more realistic disease models, paving the way for the development of a new generation of patient-on-a-chip devices. iPSCs differentiation capacity leads to invaluable improvements in personalized medicine. Moreover, the connection of single-OoC into multi-OoC or body-on-a-chip allows to investigate drug pharmacodynamic and pharmacokinetics through the study of multi-organs cross-talks. The need of a breakthrough thanks to this technology is particularly relevant within the field of neurodegenerative diseases, where the number of patients is increasing and the successful rate in drug discovery is worryingly low. In this review we discuss current iPSC-based OoC as drug screening models and their implication in development of new therapies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fanizza
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Marzia Campanile
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and
Chemical Engineering “Giulio Natta,” Politecnico di Milano, Milan, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di
Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
48
|
Gajera CR, Fernandez R, Postupna N, Montine KS, Keene CD, Bendall SC, Montine TJ. Mass Synaptometry: Applying Mass Cytometry to Single Synapse Analysis. Methods Mol Biol 2022; 2417:69-88. [PMID: 35099792 PMCID: PMC8820390 DOI: 10.1007/978-1-0716-1916-2_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Synaptic degeneration is one of the earliest and phenotypically most significant features associated with numerous neurodegenerative conditions, including Alzheimer's and Parkinson's diseases. Synaptic changes are also known to be important in neurocognitive disorders such as schizophrenia and autism spectrum disorders. Several labs, including ours, have demonstrated that conventional (fluorescence-based) flow cytometry of individual synaptosomes is a robust and reproducible method. However, the repertoire of probes needed to assess comprehensively the type of synapse, pathologic proteins (including protein products of risk genes discovered in GWAS), and markers of stress and injury far exceeds what is achievable with conventional flow cytometry. We recently developed a method that applies CyTOF (Cytometry by Time-Of-Flight mass spectrometry) to high-dimensional analysis of individual human synaptosomes, overcoming many of the multiplexing limitations of conventional flow cytometry. We call this new method Mass Synaptometry. Here we describe the preparation of synaptosomes from human and mouse brain, the generation and quality control of the "SynTOF" (Synapse by Time-Of-Flight mass spectrometry) antibody panel, the staining protocol, and CyTOF parameter setup for acquisition, post-acquisition processing, and analysis.
Collapse
Affiliation(s)
- Chandresh R. Gajera
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Rosemary Fernandez
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Kathleen S. Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, United States
| | - Sean C. Bendall
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Thomas J. Montine
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| |
Collapse
|
49
|
Chan AWS, Cho IK, Li CX, Zhang X, Patel S, Rusnak R, Raper J, Bachevalier J, Moran SP, Chi T, Cannon KH, Hunter CE, Martin RC, Xiao H, Yang SH, Gumber S, Herndon JG, Rosen RF, Hu WT, Lah JJ, Levey AI, Smith Y, Walker LC. Cerebral Aβ deposition in an Aβ-precursor protein-transgenic rhesus monkey. AGING BRAIN 2022; 2:100044. [PMID: 36589695 PMCID: PMC9802652 DOI: 10.1016/j.nbas.2022.100044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
With the ultimate goal of developing a more representative animal model of Alzheimer's disease (AD), two female amyloid-β-(Aβ) precursor protein-transgenic (APPtg) rhesus monkeys were generated by lentiviral transduction of the APP gene into rhesus oocytes, followed by in vitro fertilization and embryo transfer. The APP-transgene included the AD-associated Swedish K670N/M671L and Indiana V717F mutations (APPSWE/IND) regulated by the human polyubiquitin-C promoter. Overexpression of APP was confirmed in lymphocytes and brain tissue. Upon sacrifice at 10 years of age, one of the monkeys had developed Aβ plaques and cerebral Aβ-amyloid angiopathy in the occipital, parietal, and caudal temporal neocortices. The induction of Aβ deposition more than a decade prior to its usual emergence in the rhesus monkey supports the feasibility of creating a transgenic nonhuman primate model for mechanistic analyses and preclinical testing of treatments for Alzheimer's disease and cerebrovascular amyloidosis.
Collapse
Affiliation(s)
- Anthony W S Chan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - In Ki Cho
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Chun-Xia Li
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Xiaodong Zhang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sudeep Patel
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca Rusnak
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jessica Raper
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jocelyne Bachevalier
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Psychology, Emory College, Atlanta, GA 30322, USA
| | - Sean P Moran
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Tim Chi
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Katherine H Cannon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Carissa E Hunter
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ryan C Martin
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shang-Hsun Yang
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjeev Gumber
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - James G Herndon
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rebecca F Rosen
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lary C Walker
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
50
|
Andrews B, Murphy AE, Stofella M, Maslen S, Almeida-Souza L, Skehel JM, Skene NG, Sobott F, Frank RAW. Multidimensional dynamics of the proteome in the neurodegenerative and aging mammalian brain. Mol Cell Proteomics 2021; 21:100192. [PMID: 34979241 PMCID: PMC8816717 DOI: 10.1016/j.mcpro.2021.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 12/03/2021] [Accepted: 12/29/2021] [Indexed: 11/18/2022] Open
Abstract
The amount of any given protein in the brain is determined by the rates of its synthesis and destruction, which are regulated by different cellular mechanisms. Here, we combine metabolic labeling in live mice with global proteomic profiling to simultaneously quantify both the flux and amount of proteins in mouse models of neurodegeneration. In multiple models, protein turnover increases were associated with increasing pathology. This method distinguishes changes in protein expression mediated by synthesis from those mediated by degradation. In the AppNL-F knockin mouse model of Alzheimer’s disease, increased turnover resulted from imbalances in both synthesis and degradation, converging on proteins associated with synaptic vesicle recycling (Dnm1, Cltc, Rims1) and mitochondria (Fis1, Ndufv1). In contrast to disease models, aging in wild-type mice caused a widespread decrease in protein recycling associated with a decrease in autophagic flux. Overall, this simple multidimensional approach enables a comprehensive mapping of proteome dynamics and identifies affected proteins in mouse models of disease and other live animal test settings. Multidimensional proteomic screen to detect imbalances in mouse models of disease. Increased proteome turnover in multiple symptomatic neurodegeneration mouse models. Healthy aging is associated with a global decrease in protein turnover.
Collapse
Affiliation(s)
- Byron Andrews
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Alan E Murphy
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, W12 0BZ, UK
| | - Michele Stofella
- Astbury Centre of Molecular Structural Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK
| | - Sarah Maslen
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Leonardo Almeida-Souza
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK; Helsinki Institute of Life Science - HiLIFE, Institute of Biotechnology and Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 5, 00790, Helsinki, Finland
| | - J Mark Skehel
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Nathan G Skene
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, W12 0BZ, UK
| | - Frank Sobott
- Astbury Centre of Molecular Structural Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK
| | - René A W Frank
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK; Astbury Centre of Molecular Structural Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT, UK.
| |
Collapse
|